NCCN: Continuing Education
Target Audience: This activity is designed to meet the educational needs of physicians, nurses, and pharmacists involved in the management of patients with cancer.
Accreditation Statement
Physicians: National Comprehensive Cancer Network is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to provide continuing medical education for physicians.
NCCN designates this journal-based CE activity for a maximum of 1.0 AMA PRA Category 1 Credit™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.
Nurses: National Comprehensive Cancer Network is accredited as a provider of continuing nursing education by the American Nurses Credentialing Center‘s Commission on Accreditation.
NCCN designates this educational activity for a maximum of 1.0 contact hour.
Pharmacists: National Comprehensive Cancer Network is accredited by the Accreditation Council for Pharmacy Education as a provider of continuing pharmacy education.
NCCN designates this knowledge-based continuing education activity for 1.0 contact hour (0.1 CEUs) of continuing education credit. UAN: 0836-0000-17-003-H01-P
All clinicians completing this activity will be issued a certificate of participation. To participate in this journal CE activity: 1) review the educational content; 2) take the posttest with a 66% minimum passing score and complete the evaluation at http://education.nccn.org/node/80480; and 3) view/print certificate.
Release date: March 10, 2017; Expiration date: March 10, 2018
Learning Objectives:
Upon completion of this activity, participants will be able to:
Integrate into professional practice the updates to NCCN Guidelines for CLL/SLL
Describe the rationale behind the decision-making process for developing the NCCN Guidelines for CLL/SLL
NCCN Categories of Evidence and Consensus
Category 1: Based upon high-level evidence, there is uniform NCCN consensus that the intervention is appropriate.
Category 2A: Based upon lower-level evidence, there is uniform NCCN consensus that the intervention is appropriate.
Category 2B: Based upon lower-level evidence, there is NCCN consensus that the intervention is appropriate.
Category 3: Based upon any level of evidence, there is major NCCN disagreement that the intervention is appropriate.
All recommendations are category 2A unless otherwise noted.
Clinical trials: NCCN believes that the best management for any cancer patient is in a clinical trial. Participation in clinical trials is especially encouraged.
Overview
Chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL) constitute approximately 7% of newly diagnosed non-Hodgkin's lymphomas (NHL) cases.1 In 2016, an estimated 18,960 people were diagnosed with CLL in the United States and an estimated 4,660 people died of the disease.2 CLL/SLL is characterized by a progressive accumulation of leukemic cells in the blood, bone marrow, and lymphoid tissues.
CLL and SLL are different manifestations of the same disease and are managed in much the same way.3 The major difference between them is that a significant number of the abnormal lymphocytes are found in blood in CLL, in addition to bone marrow and lymphoid tissue; in SLL there are few, if any, abnormal lymphocytes circulating in the blood, and the bulk of disease is in the lymph nodes, bone marrow, and other lymphoid tissues.
During the past several decades, therapeutic options for CLL have evolved significantly. The advent
of immunomodulating agents (eg, lenalidomide) and CD20 (rituximab, obinutuzumab, and ofatumumab) and CD52 (alemtuzumab) monoclonal antibodies have led to the development of effective chemoimmunotherapy regimens. More recently, small molecule inhibitors targeting kinases involved in a number of critical signaling pathways (eg, Bruton tyrosine kinase [BTK], phosphatidylinositol 3-kinase [PI3K], and spleen tyrosine kinase) and a small molecule inhibitor of the BCL-2 family of proteins have shown promising activity in the treatment of patients with CLL (Tables 1 and 2).These NCCN Guidelines Insights highlight important updates to the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for CLL/SLL for the treatment of patients with newly diagnosed or relapsed or refractory CLL/SLL.
First-Line Therapy
First-line therapy with fludarabine/cyclophosphamide/rituximab (FCR) results in high response rates and improved overall survival (OS) in specific subgroups of fit patients with previously untreated CLL, especially those with mutated IGHV.4,5 In a phase II study of 300 patients with previously untreated CLL, at a median follow-up of 12.8 years, the overall response rate (ORR) was 95% (72% achieved a complete response [CR]),4 median progression-free survival (PFS) was 6.4 years, and the overall 12.8-year PFS rate was 30.9% (53.9% for those with mutated IGHV and 8.7% for unmutated IGHV). Minimal residual disease (MRD) negativity was achieved in 50.7% of patients with mutated IGHV, with a PFS rate of 79.8% at 12.8 years. In a multivariable analysis, unmutated IGHV and del(17p) by conventional karyotyping were significantly associated with inferior PFS. Long-term PFS was notable particularly for patients with mutated IGHV, with a plateau on the PFS curve beyond 10 years. In a large, international, randomized, phase III clinical trial (CLL8 study), 817 physically fit patients with previously untreated CLL (median age, 61 years) were randomized to receive
up to 6 courses of either FCR (n=408) or fludarabine/cyclophosphamide (FC; n=409).5 The FCR regimen resulted in a higher ORR (90% vs 80%; P<.001) and CR rate (44% vs 22%; P<.001) compared with FC. With a median follow-up of 5.9 years, the median PFS was 56.8 months for FCR and 33 months for FC (P<.001); median OS was not reached for FCR and was 86.0 months for FC (P=.001).5 FCR was associated with a statistically significant survival benefit compared with FC in patients aged <65 years (5-year OS, 80.9% and 69.2%, respectively; P=.002); corresponding 5-year OS rates were 73.9% and 61.6%, respectively (P=.288). The incidence of prolonged neutropenia was significantly higher with the FCR regimen than with FC during the first year after treatment (16.6% vs 8.8%; P=.007).Bendamustine, an alkylating agent with a low cross-resistance to other alkylating agents, was evaluated as first-line monotherapy and in combination with rituximab.6–8 In a multicenter phase II trial (CLL2M study), bendamustine/rituximab (BR) resulted in high ORR rates (88%; CR, 23%) in previously untreated patients (n=117; 26% were aged >70 years), with similar response and survival outcomes among the subgroup of elderly patients (aged >70 years).6 After a median follow-up of 27 months, median PFS was 34 months and the OS rate was 90.5%. Thrombocytopenia (22%), neutropenia (20%), anemia (20%), allergic/infusion reactions (9%), and infections (8%) were the most common grade 3 or 4 toxicities.
The final analysis of the CLL10 study, which compared BR with FCR as first-line therapy for CLL in fit patients without del(17p) (n=567; Cumulative illness Rating Scale [CIRS] score ≤6, creatinine clearance [CrCl] >70 mL/min), confirmed the superiority in PFS with FCR over BR in patients aged ≤65 years without significant comorbidities.7 The median age was 61.6 years for all patients, but a significantly higher proportion of patients were >65 years in the BR arm (39% vs 30%). After a median follow-up of 37.1 months, the ORR was 95% for FCR and 96%
for BR (P=1.0) with no difference in OS (3-year OS rate, 91% vs 92%, respectively; P=.89). FCR resulted in a higher CR rate (40% vs 31%), more MRD negativity (59% vs 26% at 12 months; P<.0001; and 55% vs 27% at 18 months; P=.002), and longer median PFS (55.2 vs 41.7 months; P=.0003) compared with BR. The PFS benefit of FCR was significant in physically fit patients aged <65 years and in those with mutated IGHV; the median PFS was 53.6 and 38.5 months, respectively, for FCR and BR (P=.0004). There was no significant difference in PFS between FCR and BR for patients aged >65 years (median not reached vs 48.5 months, respectively; P=.172). Among patients with mutated IGHV, the median PFS was not reached for FCR compared with 55.4 months for BR (P=.089). The incidence of severe neutropenia and infections were significantly more frequent in the FCR arm (39% vs 25%), especially in those aged >65 years.Updated results of the study confirmed that BR is also associated with a decreased risk of secondary acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS).9 After a median follow-up of 58.2 months, the incidences of secondary AML and MDS were 3% and 1% in the FCR and BR arms, respectively. The results of the study confirm that FCR remains the standard first-line therapy for untreated CLL in fit patients and particularly for those with mutated IGHV. BR is an alternative treatment option for fit elderly patients or those with previous infections.
An ongoing phase III randomized trial is comparing rituximab/chlorambucil (R-chlorambucil) and BR as first- or second-line therapy in patients with CLL who are not candidates for fludarabine-based chemoimmunotherapy due to older age or presence of comorbid conditions.8 In an interim analysis (126 evaluable patients; 58 treated with BR, 68 treated with R-chlorambucil; median age, 74 years; range, 44–91), at the median follow-up of 24 months, the ORR among the patients who received first-line therapy was 91% in the BR group (CR,
24%) and 86% (CR, 9%) in the R-chlorambucil group.8 The median PFS was significantly longer in BR group than in the R-chlorambucil group (40 vs 30 months; P=.003), but the median OS was not significantly different (44 months vs not calculable). The incidence of adverse events (AEs) was similar between treatment groups (98% for BR vs 97% for R-chlorambucil), but the incidence of grade 3 AEs was higher for BR compared with R-chlorambucil (75% and 64%, respectively).Obinutuzumab is a glycoengineered, humanized, type II antibody targeted against CD20. Results of the CLL11 study established obinutuzumab plus chlorambucil as the new standard of care for both elderly patients and those with comorbidities lacking del(17p).10,11 In this study, 781 patients with comorbid conditions (CIRS score >6, or estimated CrCl, 30–69 mL/min) were randomized to receive either chlorambucil (n=118), obinutuzumab plus chlorambucil (n=333), or rituximab plus chlorambucil (n=330).10 Combination obinutuzumab/chlorambucil and rituximab/chlorambucil resulted in a significant improvement in median PFS compared with chlorambucil alone (26.7, 16.3, and 11.1.months, respectively; P<.001).10 The survival benefit was seen in all subgroups except patients with del(17p). After the median observation time of 39.0 months, obinutuzumab/chlorambucil significantly prolonged median PFS (28.7 vs 15.7 months; P<.001), and median time to next treatment (51 vs 38.2 months; P<.0001) compared with rituximab/chlorambucil. There was also a trend toward OS benefit for obinutuzumab.11 The most frequent grade ≥3 toxicities with obinutuzumab/chlorambucil included neutropenia (35%), infusion-related reactions (21%), thrombocytopenia (11%), and infections (11%); neutropenia (28%) and infections (14%) were the most frequent grade ≥3 toxicities associated with rituximab/chlorambucil.
Ofatumumab (a fully human anti-CD20 monoclonal antibody) was evaluated in a multicenter, open-label, phase III study (COMPLEMENT 1) as first-line treatment for patients with untreated CLL
who were considered inappropriate for fludarabine-based regimens due to advanced age and/or comorbidities; 447 patients were randomized to ofatumumab/chlorambucil versus chlorambucil monotherapy.12 After a median follow-up of 29 months, the median PFS was significantly longer for ofatumumab/chlorambucil compared with chlorambucil monotherapy (22.4 vs 13.1 months; P<.001)12; median OS was not reached in either arm. Ofatumumab/chlorambucil also resulted in a higher ORR (82% vs 69%; P=.001) and superior CR rate (12% vs 1%) compared with chlorambucil alone.Ibrutinib, an irreversible inhibitor of BTK initially approved for relapsed or refractory CLL, was also evaluated in patients with untreated CLL/SLL, including those with del(17p).13–17 Five-year follow-up data from the multicenter phase Ib/II study, which included 31 patients aged ≥65 years with untreated CLL/SLL, confirmed that ibrutinib induces very durable responses across all subgroups, including those aged ≥65 years with untreated CLL/SLL and those with del(17p), del(11q), or unmutated IGHV.14 After a median follow-up of 5 years, ibrutinib (420 mg) resulted in an ORR of 84% (CR, 29%); median PFS was not reached and the estimated PFS rate at 60 months was 92%. Responses were independent of the presence of high-risk features; however, the number of patients with del(17p), del(11q), trisomy 12, or elevated beta-2-microglobulin was small in this study. In another phase II trial that included 35 treatment-naïve patients with del(17p) treated with ibrutinib (aged ≥18 years; median, 62 years), at a median follow-up of 24 months, ibrutinib resulted in objective responses in 32 of 33 evaluable patients (55% experienced partial response [PR], and 42% experienced a PR with lymphocytosis) and the estimated OS at 24 months was 84%.15 The cumulative incidence of progression at 24 months was 9%. Grade ≥3 neutropenia, anemia, and thrombocytopenia were reported in 24%, 14%, and 10% of patients, respectively;
grade 3 pneumonia (6%) and rash (2%) were also reported.The efficacy and safety of ibrutinib in patients aged ≥65 years with untreated CLL/SLL without del(17p) was demonstrated in a randomized phase III study (RESONATE-2; 269 patients were randomized to receive ibrutinib or chlorambucil as first-line therapy).16 After a median follow-up of 28.6 months, ibrutinib resulted in a significantly higher ORR (92% vs 36%; P<.0001) and significantly longer PFS (89% vs 34% at 24 months; P<.0001) compared with chlorambucil. With 41% of patients switching to ibrutinib, the estimated 2-year OS rates in the intent-to-treat population were 95% and 84% for patients treated with ibrutinib and chlorambucil, respectively.17 Ibrutinib was approved as first-line therapy for all patients, although the efficacy of ibrutinib as first-line therapy in patients aged <65 years without del(17p) has not been established in a randomized clinical trial.
Relapsed or Refractory Therapy
The current standards of care for relapsed or refractory CLL/SLL are ibrutinib, idelalisib, and idelalisib with rituximab and venetoclax for relapsed/refractory del(17p) CLL/SLL (see Table 2).
Ibrutinib has remarkable monotherapy activity with a favorable toxicity profile in patients with relapsed or refractory CLL.18–20 In the phase III randomized RESONATE study, 391 patients with previously treated CLL were randomized to monotherapy with ibrutinib (420 mg/d) or ofatumumab.18 Most patients had advanced-stage disease and high-risk features, including del(17p), del(11q), or beta-2-microglobulin (>3.5 mg/L). At a median follow-up of 9.4 months, the ibrutinib group experienced significantly prolonged PFS (median not reached vs 8.1 months for ofatumumab) and OS (HR for death in the ibrutinib group, 0.43; P=.005; 57% reduction in the risk of death). Among patients with del(17p), median PFS was not reached for ibrutinib compared with a median PFS of 5.8 months for ofatumumab.18
At 12 months, the OS rate was 91% and 81%, respectively, for ibrutinib and ofatumumab; the ORR was also significantly higher with ibrutinib (42% vs 4%; P<.001). The most frequent nonhematologic AEs were mild (grades 1/2) with diarrhea, fatigue, pyrexia, and nausea in the ibrutinib group, and fatigue, infusion-related reactions, and cough in the ofatumumab group. Updated results of this study also confirmed that ibrutinib significantly improved ORR, PFS, and OS compared with ofatumumab in patients with CLL/SLL who had received ≥1 prior treatment.19 With a median follow-up of 16 months, the ORR (90% vs 25%; P<.0001), median PFS (not reached vs 8.1 months for ofatumumab; P<.0001), and OS rates (18-month OS, 85% and 78%, respectively) were significantly improved for ibrutinib. Results of the phase II RESONATE-17 study further confirmed the safety and efficacy of ibrutinib in 144 patients with relapsed or refractory CLL with del(17p).20 In an extended analysis with a median follow-up of 27.6 months, the investigator-assessed ORR and 24-month PFS and OS rates were 83%, 63%, and 75%, respectively.20Idelalisib (the isoform-selective oral inhibitor of PI3K-delta) has demonstrated promising clinical activity in phase I–II studies in patients with relapsed or refractory CLL/SLL, both as monotherapy and in combination with rituximab.21–23 In the multicenter phase III randomized study, 220 patients with relapsed CLL were randomized to receive rituximab with either idelalisib (150 mg) or placebo.21 Most of the patients (78%) were aged ≥65 years, 40% had moderate renal dysfunction (CrCl, <60 mL/min), 35% had poor bone marrow function (grade ≥3 cytopenias), and 85% had a CIRS score >6. At the first planned interim analysis, the study was stopped early because of the overwhelming efficacy of idelalisib plus rituximab.21 At 24 weeks, the PFS rate was 93% and 46% in the idelalisib and placebo groups, respectively. Among patients with relapsed CLL with coexisting conditions, combination idelalisib/rituximab significantly improved ORR (81% vs 13%;
P<.001), PFS (not reached in the idelalisib group vs 5.5 months for placebo), and OS at 12 months (92% vs 80%; P=.02) compared with rituximab plus placebo. Grade 3 or 4 AEs (pneumonia, pyrexia, and febrile neutropenia) were reported in 40% of patients in the idelalisib group and 35% in the placebo group. The second interim analysis of this study also confirmed the superior safety and efficacy of idelalisib plus rituximab in terms of ORR, PFS, and OS.24 The combination also retained efficacy in patients with high-risk features, such as del(17p) or TP53 mutations; unmutated IGHV, ZAP70, and CD38 expression; and beta-2-microglobulin (>4 mg/L).22 A post hoc analysis of 39 patients with relapsed or refractory SLL enrolled in phase I (n=11) and II (n=28) studies (that evaluated the efficacy and safety of patients on idelalisib with relapsed or refractory indolent NHL) showed that idelalisib monotherapy has substantial clinical activity in the subset of patients with relapsed or refractory SLL23: the ORR was 55% (6 of 11) and 61% (17 of 28); median duration of response was 2.3 and 12.5 months; and median PFS was 3.7 and 11.4 months, respectively. Idelalisib monotherapy is approved for the treatment of relapsed SLL in patients who have received ≥2 prior systemic therapies.Idelalisib in combination with rituximab is approved for the treatment of relapsed CLL in patients for whom rituximab monotherapy would be considered appropriate due to other comorbidities (reduced renal function as measured by CrCl <60 mL/min, or NCI CTCAE grade ≥3 neutropenia or thrombocytopenia resulting from myelotoxic effects of prior treatment with cytotoxic agents). Due to infection-related toxicity and deaths seen in phase III clinical trials with idelalisib in previously untreated CLL, it should not be used in the first line. Prescribers should be aware of this increased risk for infections in patients with relapsed or refractory CLL. Infection prophylaxis for herpes simplex virus, Pneumocystis jirovecii pneumonia (PJP), and routine monitoring for cytomegalovirus (CMV) reactivation are recommended for patients on idelalisib.
Venetoclax, a small-molecule BCL2 inhibitor that induces CLL cell apoptosis, has demonstrated activity in relapsed or refractory CLL.25,26 In a phase II study of 107 patients (61 patients aged ≥65 years; 46 patients aged <65 years) with relapsed or refractory del(17p) CLL, at a median follow-up of 12.1 months, venetoclax resulted in an ORR of 79.4% as assessed by the independent review committee.25 Venetoclax resulted in a high ORR (>70%) in all subgroups of patients with additional risk features [eg, fludarabine-refractory status, bulky disease, del(17p), TP53 mutation]; estimated 12-month PFS and OS rates were 72% and 86.7%, respectively. Neutropenia (40%), infection (20%), anemia (18%), and thrombocytopenia (15%) were the most common treatment-related AEs. Venetoclax has also shown promising activity in patients with relapsed or refractory CLL who have failed to respond or are intolerant to ibrutinib or idelalisib.26 In a phase II study of 64 patients with relapsed or refractory CLL after prior treatment with ibrutinib (arm A; n=43; 39 patients refractory to ibrutinib) or idelalisib (arm B; n=21; 14 patients refractory to idelalisib), venetoclax resulted in an ORR of 70% (arm A) and 48% (arm B).26 In the subset of patients with CLL refractory to ibrutinib or idelalisib, the ORR was 67% (26 of 39, arm A) and 57% (8 of 14, arm B), respectively. The estimated 12-month PFS and OS rates for all patients were 72% and 90%, respectively; median PFS and OS was not reached at the time of analysis. Neutropenia can be a longer-term toxicity requiring growth factor support and/or venetoclax dose adjustment. Initiation at a lower dose (20 mg for 1 week) and a gradual stepwise ramp-up over 5 weeks to the target dose (400 mg/d) along with prophylaxis for tumor lysis syndrome (TLS) is recommended to mitigate the risk and frequency of TLS in patients receiving venetoclax (see CSLL-G; page 304).25,27
Ofatumumab is approved for the treatment of patients with CLL refractory to fludarabine and alemtuzumab. However, it has demonstrated activity in patients with CLL refractory to fludarabine and
Treatment Options for First-line Therapy
The phase III randomized trial PROLONG evaluated the efficacy and safety of ofatumumab maintenance versus observation in 474 patients with relapsed CLL in CR or PR after second-line or third-line therapy.31 At a median follow-up of 19.1 months, ofatumumab maintenance resulted in improved PFS (29.4 vs 15.2 months, respectively; P<.0001). Neutropenia (24%) and infections (13%) were the most common grade ≥3 AEs associated with ofatumumab maintenance. Ofatumumab maintenance is approved for patients with recurrent or progressive
Treatment Options for Relapsed/Refractory Therapy
Assessment of Functional Status and Comorbidity
CLL/SLL is diagnosed mainly in older adults, with a median age at diagnosis of 72 years; approximately 70% of patients are diagnosed at age ≥65 years and 40% at age ≥75 years.32 Comorbidities are frequently present in older patients. In addition, organ function and bone marrow reserve decline with increasing age. CIRS, Charlson comorbidity index, and NCI comorbidity index are some of the scoring systems that can be used to assess comorbidities in patients with CLL. CIRS in combination with CrCl was used by the German CLL Study Group (GCLLSG) to assess the overall fitness of patients enrolled in clinical trials.33,34 In a study that assessed the comorbidity burden and investigated its impact on treatment in 555 patients with untreated CLL enrolled in 2 GCLLSG trials, 26% of patients had comorbidities involving the metabolic/endocrine system, 21% had comorbidities in vascular system, and 12% had cardiac comorbidities.33 The presence of multiple comorbidities (≥2 comorbidities) was an independent predictor of clinical outcome independent of patients' age or disease stage.33 The median OS (71.7 vs 90.2 months; P<.001) and PFS (21.0 vs 31.5 months; P<.01) were significantly shorter for patients with ≥2 comorbidities than for those with <2 comorbidities. These findings underscore the need to assess comorbidities, in addition to patient age and performance status (PS), before treatment selection. Older patients with adequate functional status can be treated with more active or intensive therapies,35 and should be evaluated for cytogenetic abnormalities using fluorescence in situ hybridization. Patient age and the presence or absence of del(17p) or TP53 mutations should then help direct treatment options, as discussed in the following section.
Treatment Recommendations
Early-stage disease in some patients may have an indolent course and in others may progress rapidly to advanced disease requiring immediate treatment. Patients with stage II–IV SLL, low-risk CLL (Rai stage 0; Binet A), or intermediate-risk CLL (Rai stage I–II; Binet B) may benefit from treatment if they become symptomatic or show evidence of progressive disease.36 Patients with advanced-stage or high-risk CLL (Rai stage III–IV; Binet C) with progressive cytopenia require treatment (see CSLL-3; page 295. Selected patients with mild, stable cytopenia may continue to be observed.
Absolute lymphocyte count alone is not an indication for treatment unless it is >200 to 300×109/L or symptoms related to leukostasis occur.36 Indications for initiating treatment include severe fatigue, weight loss, night sweats, and fever without infection; threatened end-organ function; progressive bulky disease (enlarged spleen or lymph nodes); progressive anemia or thrombocytopenia; or autoimmune anemia/thrombocytopenia unresponsive to corticosteroids.36 Asymptomatic patients should be observed until these indications become apparent. Recommendations for patients with indications for treatment are based on patient's age or functional status (comorbidity index/PS), as well as the presence or absence of del(17p) or TP53 mutations.
The age cutoff of 65 years is used in most clinical trials, including those conducted by the GCLLSG. In a retrospective analysis that evaluated the impact of age on outcome after initial treatment with different chemoimmunotherapy and chemotherapy regimens in patients with CLL enrolled in CALGB trials, the benefit of fludarabine compared with chlorambucil decreased marginally with age, with estimated hazard ratios of 0.70, 0.76, and 0.81 at 65, 70, and 75 years, respectively.37 The benefit of fludarabine relative to chlorambucil also decreased at an earlier age for OS than for PFS, with the estimated hazard ratios of 0.88, 1.01, and 1.15 at 65, 70, and 75 years, respectively. In addition, approximately 44% of patients aged >65 years have some degree of chronic kidney disease, which also increases the likelihood of toxicity associated with fludarabine-based regimens.38 Based on these data, the panel decided to change the age cutoff from 70 to 65 years.
CLL Without del(17p) or TP53 Mutations
First-Line Therapy: Obinutuzumab/chlorambucil (category 1),10,11 ibrutinib (category 1),16 and ofatumumab/chlorambucil12 are the preferred treatment options for frail patients with significant comorbidities (eg, not able to tolerate purine analogs) or patients aged ≥65 years and younger patients with significant comorbidities (see CSLL-5; page 297). Bendamustine with or without rituximab is a reasonable alternative for patients aged ≥65 years who are otherwise eligible for chemoimmunotherapy.6,7 In the CLL11 trial, there was a clinically meaningful improvement in PFS with a trend toward improved OS in patients treated with obinutuzumab/chlorambucil versus rituximab/chlorambucil.10,11 Therefore, rituximab/chlorambucil should be reserved for patients who cannot tolerate obinutuzumab.39,40
With multiple randomized studies showing a survival advantage for combination regimens containing chlorambucil or rituximab compared with monotherapy with either of these agents,10,12 most of the panel members acknowledged that monotherapy with chlorambucil or rituximab is not an effective first-line treatment even for frail patients with comorbid conditions. However, some panel members felt that given the favorable tolerability profile, monotherapy with rituximab or chlorambucil may be an appropriate treatment option for a small fraction of very frail patients or patients aged ≥65 years with substantial comorbidities or decreased PS for whom more intensive regimens are not appropriate.41,42 Rituximab and chlorambucil are included as alternate treatment options (category 3).
In patients aged <65 years without significant comorbidities, fludarabine-based chemoimmunotherapy has emerged as the standard of care, especially in those with mutated IGHV.4,5 Therefore, chemoimmunotherapy with FCR is the preferred treatment option (category 1) for these patients. FR, pentostatin/cyclophosphamide/rituximab (PCR), or bendamustine with or without rituximab are included as alternate options for chemoimmunotherapy.
The panel acknowledged that there are no data to support the inclusion of ibrutinib as a category 1 recommendation for patients aged <65 years, because the RESONATE-2 trial (on which the FDA based its approval of ibrutinib for first-line therapy in all patients with CLL/SLL) established the efficacy of ibrutinib as first-line therapy only in patients aged ≥65 years without del(17p).16 However, with the recent FDA approval, some panel members agreed that ibrutinib may be an appropriate option instead of chemoimmunotherapy in younger patients with unmutated IGHV who do want to enroll in a clinical trial. Therefore, the panel included ibrutinib as a category 2A recommendation for first-line therapy for patients aged <65 years without del(17p).
Relapsed or Refractory Therapy: Ibrutinib (category 1),18,19 idelalisib/rituximab (category 1),21 idelalisib,23 and venetoclax (particularly for patients deemed intolerant or refractory to ibrutinib or idelalisib)26 are included as preferred options for patients with relapsed or refractory disease, regardless of their age and comorbidities. The results of recent phase III trials have shown that the addition of idelalisib or ibrutinib to BR significantly improves PFS in patients with relapsed or refractory CLL.43,44 BR with idelalisib or ibrutinib is included an alternate option regardless of patient age and comorbidities.
For frail patients with significant comorbidities or patients aged ≥65 years and younger patients with comorbidities, reduced-dose FCR or PCR, bendamustine with or without rituximab, high-dose methylprednisolone (HDMP) or chlorambucil with rituximab, monotherapy with ofatumumab or obinutuzumab, lenalidomide or alemtuzumab with or without rituximab, or dose-dense rituximab are included as alternative options. For patients aged <65 years without significant comorbidities, chemoimmunotherapy (FCR, FC + ofatumumab, PCR, bendamustine ± rituximab, CHOP + rituximab, OFAR [oxaliplatin/fludarabine/cytarabine/rituximab]), monotherapy with ofatumumab or obinutuzumab, lenalidomide or alemtuzumab with or without rituximab, or HDMP with rituximab are included as alternate options.
Ofatumumab maintenance (category 2B) is recommended for patients who are in CR or PR as second-line therapy.31 Allogeneic hematopoietic cell transplant (HCT) can be considered for select patients (without significant comorbidities) after reinduction of remission.
CLL With del(17p) or TP53 Mutations
Enrollment in an appropriate clinical trial is recommended for patients with del(17p). In the absence of appropriate clinical trials, ibrutinib is the preferred treatment option for first-line therapy13,15; HDMP plus rituximab,45,46 obinutuzumab/chlorambucil,10 and alemtuzumab with or without rituximab47–49 are included as alternate treatment options (see CSLL-D, 3 of 5; page 301). The efficacy of ibrutinib in del(17p) CLL/SLL exceeds the results of alternative regimens in the upfront and salvage settings, and should be considered the best choice in the absence of a contraindication.
Recent reports suggest that complex karyotype (≥3 unrelated chromosomal abnormalities in >1 cell on CpG-stimulated karyotype of CLL cells) is a stronger predictor of poor clinical outcomes than del(17p) in patients with relapsed or refractory CLL treated with ibrutinib-based regimens.50,51 In an analysis of 88 patients with relapsed or refractory CLL treated with ibrutinib-based regimens, in a multivariate analysis, only complex karyotype was significantly associated with inferior event-free survival (P=.006), whereas complex karyotype (P=.008) and fludarabine-refractory CLL (P=.005) were independently associated with inferior OS.51 Complex karyotype has also been identified as an independent prognostic factor for shorter time to first treatment.52 CpG-stimulated karyotyping is useful for identifying high-risk patients, particularly for treatment with ibrutinib.
Patients with response to first-line therapy should be considered for allogeneic HCT, if complex karyotype is present. However, available data suggest that complex karyotype (≥5 abnormalities) is associated with inferior OS and event-free survival after allogeneic SCT with reduced-intensity conditioning in patients with high-risk interphase cytogenetics.53 Allogeneic HCT is not recommended for patients with del(17p) CLL/SLL responding to first-line therapy with ibrutinib, if complex karyotype is not present. Ofatumumab maintenance (category 2B) is recommended for patients experiencing CR or PR to second-line therapy.31
Patients with no response to first-line therapy should be enrolled in clinical trials or be treated with second-line therapy for relapsed or refractory disease; preferred options for relapsed or refractory disease are ibrutinib,18–20 venetoclax,25 idelalisib/rituximab,22 and idelalisib.23
Special Considerations for the Use of Small Molecule Inhibitors
Ibrutinib and idelalisib cause early mobilization of lymphocytes into the blood, resulting in a transient increase in absolute lymphocyte count in most patients, which does not signify disease progression.54,55 This onset of isolated lymphocytosis occurs during the first few weeks of ibrutinib treatment and may persist for several weeks on treatment. Although lymphocytosis can sometimes be profound, clinical consequence (ie, leukostasis) is extremely rare and treatment should be continued. Slow or incomplete resolution of lymphocytosis does not appear to impact outcome as measured by PFS (CSLL-F; pages 302 and 303).54
Atrial fibrillation (grade ≥3) and major hemorrhage (defined as serious or grade ≥3 bleeding events or central nervous system hemorrhage of any grade) have been reported in 6% and 4% of patients treated with ibrutinib, respectively.13,16 The benefit and risk of ibrutinib should be evaluated in patients requiring antiplatelet or anticoagulant therapies. Patients requiring warfarin have been excluded from clinical trials evaluating ibrutinib, and should not be given concomitantly with warfarin. Hypertension (grade ≥3) associated with ibrutinib (reported in 20%) has uncommonly been the basis for discontinuation and should be managed with antihypertensives as appropriate.13 Alternate treatment should be considered, especially in patients with hypertension that is not medically controllable. At the time of disease progression on ibrutinib, transition to alternate treatment should be performed as soon as possible upon stopping ibrutinib, because progression may accelerate when ibrutinib is stopped. Treatment-free interval should be as short as possible (CSLL-F; pages 302 and 303).
Fatal and/or serious hepatotoxicity, severe diarrhea or colitis, pneumonitis, and intestinal perforation have been observed in patients treated with idelalisib.56 Hepatotoxicity is a major concern in younger patients treated with idelalisib as first-line therapy.57 Close monitoring of transaminase levels is essential, and concurrent administration of idelalisib with other hepatotoxic drugs should be avoided. Idelalisib is also associated with opportunistic infections and febrile neutropenia. PJP and CMV reactivation have been reported in patients treated with idelalisib, and the risk of developing these opportunistic infections is highest in the first 6 months of treatment.43 The addition of CD20 monoclonal antibody or chemoimmunotherapy to idelalisib increases the risk of febrile neutropenia. Appropriate anti-infective prophylaxis and routine monitoring for early signs of infectious complications and CMV reactivation is recommended for patients receiving idelalisib (CSLL-F; pages 302 and 303).
Conclusions
The choice of first-line treatment for CLL/SLL should be based on disease stage, presence or absence of del(17p) or TPwent. Ibrutinib, idelalisib (± rituximab), and venetoclax are effective treatment options for relapsed or refractory del(17p) CLL/SLL. Careful monitoring of AEs after treatment initiation and supportive care for treatment-related complications should be an integral part of management of patients with CLL/SLL.
References
- 1.↑
A clinical evaluation of the international lymphoma study group classification of non-Hodgkin's lymphoma. The non-Hodgkin's lymphoma classification project. Blood 1997;89:3909–3918.
- 3.↑
Tsimberidou AM, Wen S, O'Brien S et al.. Assessment of chronic lymphocytic leukemia and small lymphocytic lymphoma by absolute lymphocyte counts in 2,126 patients: 20 years of experience at the University of Texas M.D. Anderson Cancer Center. J Clin Oncol 2007;25:4648–4656.
- 4.↑
Thompson PA, Tam CS, O'Brien SM et al.. Fludarabine, cyclophosphamide, and rituximab treatment achieves long-term disease-free survival in IGHV-mutated chronic lymphocytic leukemia. Blood 2015;127:303–309.
- 5.↑
Fischer K, Bahlo J, Fink AM et al.. Long-term remissions after FCR chemoimmunotherapy in previously untreated patients with CLL: updated results of the CLL8 trial. Blood 2016;127:208–215.
- 6.↑
Fischer K, Cramer P, Busch R et al.. Bendamustine in combination with rituximab for previously untreated patients with chronic lymphocytic leukemia: a multicenter phase II trial of the German Chronic Lymphocytic Leukemia Study Group. J Clin Oncol 2012;30:3209–3216.
- 7.↑
Eichhorst B, Fink AM, Bahlo J et al.. First-line chemoimmunotherapy with bendamustine and rituximab versus fludarabine, cyclophosphamide, and rituximab in patients with advanced chronic lymphocytic leukaemia (CLL10): an international, open-label, randomised, phase 3, non-inferiority trial. Lancet Oncol 2016;17:928–942.
- 8.↑
Michallet AS, Aktan M, Schuh A. Rituximab in combination with bendamustine or chlorambucil for the treatment of chronic lymphocytic leukemia: primary results from the randomized phase IIIb MaBLe study. Abstract 178. Available at: https://static1.squarespace.com/static/52f8f139e4b0bae912c96d0d/t/562a508ae4b05e1ace338235/1445613706834/IWCLL+Report+_v4+.pdf. Accessed February 13, 2017.
- 9.↑
Eichhorst BF, Bahlo J, Maurer C et al.. Favorable toxicity profile and long term outcome of elderly, but physically fit CLL patients (pts) receiving first line bendamustine and rituximab (BR) frontline chemoimmunotherapy in comparison to fludarabine, cyclophosphamide, and rituximab (FCR) in advanced chronic lymphocytic leukemia (CLL): update analysis of an international, randomized study of the German CLL Study Group (GCLLSG) (CLL10 study) [abstract]. Blood 2016;128:Abstract 4382.
- 10.↑
Goede V, Fischer K, Busch R et al.. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med 2014;370:1101–1111.
- 11.↑
Goede V, Fischer K, Bosch F et al.. Updated survival analysis from the CLL11 study: obinutuzumab versus rituximab in chemoimmunotherapy-treated patients with chronic lymphocytic leukemia [abstract]. Blood 2015;126:Abstract 1733.
- 12.↑
Hillmen P, Robak T, Janssens A et al.. Chlorambucil plus ofatumumab versus chlorambucil alone in previously untreated patients with chronic lymphocytic leukaemia (COMPLEMENT 1): a randomised, multicentre, open-label phase 3 trial. Lancet 2015;385:1873–1883.
- 13.↑
Byrd JC, Furman RR, Coutre SE et al.. Three-year follow-up of treatment-naive and previously treated patients with CLL and SLL receiving single-agent ibrutinib. Blood 2015;125:2497–2506.
- 14.↑
O'Brien SM, Furman RR, Coutre SE et al.. Five-year experience with single-agent ibrutinib in patients with previously untreated and relapsed/refractory chronic lymphocytic leukemia/small lymphocytic leukemia [abstract]. Blood 2016;128:Abstract 233.
- 15.↑
Farooqui MZ, Valdez J, Martyr S et al.. Ibrutinib for previously untreated and relapsed or refractory chronic lymphocytic leukaemia with TP53 aberrations: a phase 2, single-arm trial. Lancet Oncol 2015;16:169–176.
- 16.↑
Burger JA, Tedeschi A, Barr PM et al.. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med 2015;373:2425–2437.
- 17.↑
Barr P, Robak T, Owen CJ et al.. Updated efficacy and safety from the phase 3 RESONATE-2 study: ibrutinib as first-line treatment option in patients 65 years and older with chronic lymphocytic leukemia/small lymphocytic leukemia [abstract]. Blood 2016;128:Abstract 234.
- 18.↑
Byrd JC, Brown JR, O'Brien S et al.. Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. N Engl J Med 2014;371:213–223.
- 19.↑
Brown JR, Hillmen P, O'Brien S et al.. Updated efficacy including genetic and clinical subgroup analysis and overall safety in the phase 3 RESONATE trial of ibrutinib versus ofatumumab in previously treated chronic lymphocytic leukemia/small lymphocytic lymphoma [abstract]. Blood 2014;124:Abstract 3331.
- 20.↑
O'Brien S, Jones JA, Coutre SE et al.. Ibrutinib for patients with relapsed or refractory chronic lymphocytic leukaemia with 17p deletion (RESONATE-17): a phase 2, open-label, multicentre study. Lancet Oncol 2016;17:1409–1418.
- 21.↑
Furman RR, Sharman JP, Coutre SE et al.. Idelalisib and rituximab in relapsed chronic lymphocytic leukemia. N Engl J Med 2014;370:997–1007.
- 22.↑
Sharman JP, Coutre SE, Furman RR et al.. Efficacy of idelalisib in CLL subpopulations harboring del(17p) and other adverse prognostic factors: results from a phase 3, randomized, double-blind, placebo-controlled trial [abstract]. J Clin Oncol 2014;32(15 Suppl):Abstract 7011.
- 23.↑
Gopal AK, Davies AJ, Flinn IW et al.. Idelalisib monotherapy and durable responses in patients with relapsed or refractory small lymphocytic lymphoma (SLL) [abstract]. Blood 2015;126:Abstract 2743.
- 24.↑
Sharman JP, Coutre SE, Furman RR et al.. Second interim analysis of a phase 3 study of idelalisib (Zydelig) plus rituximab (R) for relapsed chronic lymphocytic leukemia (CLL): efficacy analysis in patient subpopulations with del(17p) and other adverse prognostic factors [abstract]. Blood 2014;124:Abstract 330.
- 25.↑
Stilgenbauer S, Eichhorst B, Schetelig J et al.. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: a multicentre, open-label, phase 2 study. Lancet Oncol 2016;17:768–778.
- 26.↑
Jones J, Choi MY, Mato AR et al.. Venetoclax (VEN) monotherapy for patients with chronic lymphocytic leukemia (CLL) who relapsed after or were refractory to ibrutinib or idelalisib [abstract]. Blood 2016;128:Abstract 637.
- 27.↑
Seymour JF. Effective mitigation of tumor lysis syndrome with gradual venetoclax dose ramp, prophylaxis, and monitoring in patients with chronic lymphocytic leukemia. Ann Hematol 2016;95:1361–1362.
- 28.↑
Wierda WG, Padmanabhan S, Chan GW et al.. Ofatumumab is active in patients with fludarabine-refractory CLL irrespective of prior rituximab: results from the phase II international study. Blood 2011;118:5126–5129.
- 29.↑
Osterborg A, Jewell RC, Padmanabhan-Iyer S et al.. Ofatumumab monotherapy in fludarabine-refractory chronic lymphocytic leukemia: final results from a pivotal study. Haematologica 2015;100:e311–314.
- 30.↑
Robak T, Warzocha K, Govind Babu K et al.. Ofatumumab plus fludarabine and cyclophosphamide in relapsed chronic lymphocytic leukemia: results from the COMPLEMENT 2 trial. Leuk Lymphoma 2017;58:1084–1093.
- 31.↑
van Oers MH, Kuliczkowski K, Smolej L et al.. Ofatumumab maintenance versus observation in relapsed chronic lymphocytic leukaemia (PROLONG): an open-label, multicentre, randomised phase 3 study. Lancet Oncol 2015;16:1370–1379.
- 32.↑
SEER Stat Facts: Chronic Lymphocytic Leukemia (CLL). National Cancer Institute Web site. Available at: http://seer.cancer.gov/statfacts/html/clyl.html. Accessed August 2012.
- 33.↑
Goede V, Cramer P, Busch R et al.. Interactions between comorbidity and treatment of chronic lymphocytic leukemia: results of German Chronic Lymphocytic Leukemia Study Group trials. Haematologica 2014;99:1095–1100.
- 34.↑
Salvi F, Miller MD, Grilli A et al.. A manual of guidelines to score the modified cumulative illness rating scale and its validation in acute hospitalized elderly patients. J Am Geriatr Soc 2008;56:1926–1931.
- 35.↑
Satram-Hoang S, Reyes C, Hoang KQ et al.. Treatment practice in the elderly patient with chronic lymphocytic leukemia-analysis of the combined seer and medicare database. Ann Hematol 2014;93:1335–1344.
- 36.↑
Hallek M, Cheson BD, Catovsky D et al.. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood 2008;111:5446–5456.
- 37.↑
Woyach JA, Ruppert AS, Rai K et al.. Impact of age on outcomes after initial therapy with chemotherapy and different chemoimmunotherapy regimens in patients with chronic lymphocytic leukemia: results of sequential cancer and leukemia group b studies. J Clin Oncol 2013;31:440–447.
- 38.↑
Stevens LA, Li S, Wang C et al.. Prevalence of CKD and comorbid illness in elderly patients in the United States: results from the Kidney Early Evaluation Program (KEEP). Am J Kidney Dis 2010;55:S23–33.
- 39.↑
Hillmen P, Gribben JG, Follows GA et al.. Rituximab plus chlorambucil as first-line treatment for chronic lymphocytic leukemia: final analysis of an open-label phase II study. J Clin Oncol 2014;32:1236–1241.
- 40.↑
Foa R, Giudice ID, Cuneo A et al.. Chlorambucil plus rituximab with or without maintenance rituximab as first-line treatment for elderly chronic lymphocytic leukemia patients. Am J Hematol 2014;89:480–486.
- 41.↑
Hainsworth JD, Litchy S, Barton JH et al.. Single-agent rituximab as first-line and maintenance treatment for patients with chronic lymphocytic leukemia or small lymphocytic lymphoma: a phase II trial of the Minnie Pearl Cancer Research Network. J Clin Oncol 2003;21:1746–1751.
- 42.↑
Eichhorst BF, Busch R, Stilgenbauer S et al.. First-line therapy with fludarabine compared with chlorambucil does not result in a major benefit for elderly patients with advanced chronic lymphocytic leukemia. Blood 2009;114:3382–3391.
- 43.↑
Zelenetz AD, Brown JR, Delgado J et al.. Updated analysis of overall survival in randomized phase III study of idelalisib in combination with bendamustine and rituximab in patients with relapsed/refractory CLL [abstract]. Blood 2016;128:Abstract 231.
- 44.↑
Chanan-Khan A, Cramer P, Demirkan F et al.. Ibrutinib combined with bendamustine and rituximab compared with placebo, bendamustine, and rituximab for previously treated chronic lymphocytic leukaemia or small lymphocytic lymphoma (helios): a randomised, double-blind, phase 3 study. Lancet Oncol 2016;17:200–211.
- 45.↑
Bowen DA, Call TG, Jenkins GD et al.. Methylprednisolone-rituximab is an effective salvage therapy for patients with relapsed chronic lymphocytic leukemia including those with unfavorable cytogenetic features. Leuk Lymphoma 2007;48:2412–2417.
- 46.↑
Castro JE, Sandoval-Sus JD, Bole J et al.. Rituximab in combination with high-dose methylprednisolone for the treatment of fludarabine refractory high-risk chronic lymphocytic leukemia. Leukemia 2008;22:2048–2053.
- 47.↑
Keating MJ, Flinn I, Jain V et al.. Therapeutic role of alemtuzumab (Campath-1H) in patients who have failed fludarabine: results of a large international study. Blood 2002;99:3554–3561.
- 48.
Lozanski G, Heerema NA, Flinn IW et al.. Alemtuzumab is an effective therapy for chronic lymphocytic leukemia with p53 mutations and deletions. Blood 2004;103:3278–3281.
- 49.↑
Fiegl M, Stauder R, Steurer M et al.. Alemtuzumab in chronic lymphocytic leukemia: final results of a large observational multicenter study in mostly pretreated patients. Ann Hematol 2014;93:267–277.
- 50.↑
Woyach JA, Ruppert AS, Lozanski G et al.. Association of disease progression on ibrutinib therapy with the acquisition of resistance mutations: a single-center experience of 267 patients [abstract]. J Clin Oncol 2014;32(Suppl):Abstract 7010.
- 51.↑
Thompson PA, O'Brien SM, Wierda WG et al.. Complex karyotype is a stronger predictor than del(17p) for an inferior outcome in relapsed or refractory chronic lymphocytic leukemia patients treated with ibrutinib-based regimens. Cancer 2015;121:3612–3621.
- 52.↑
Baliakas P, Iskas M, Gardiner A et al.. Chromosomal translocations and karyotype complexity in chronic lymphocytic leukemia: a systematic reappraisal of classic cytogenetic data. Am J Hematol 2014;89:249–255.
- 53.↑
Jaglowski SM, Ruppert AS, Heerema NA et al.. Complex karyotype predicts for inferior outcomes following reduced-intensity conditioning allogeneic transplant for chronic lymphocytic leukaemia. Br J Haematol 2012;159:82–87.
- 54.↑
Woyach JA, Smucker K, Smith LL et al.. Prolonged lymphocytosis during ibrutinib therapy is associated with distinct molecular characteristics and does not indicate a suboptimal response to therapy. Blood 2014;123:1810–1817.
- 55.↑
Brown JR, Byrd JC, Coutre SE et al.. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110delta, for relapsed/refractory chronic lymphocytic leukemia. Blood 2014;123:3390–3397.
- 56.↑
Coutre SE, Barrientos JC, Brown JR et al.. Management of adverse events associated with idelalisib treatment: expert panel opinion. Leuk Lymphoma 2015;56:2779–2786.
- 57.↑
Lampson BL, Kasar SN, Matos TR et al.. Idelalisib given front-line for treatment of chronic lymphocytic leukemia causes frequent immune-mediated hepatotoxicity. Blood 2016;128:195–203.